Cargando…

Recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the SARS-CoV-2 virus

BACKGROUND: Plasma gelsolin (pGSN) is an important part of the blood actin buffer that prevents negative consequences of possible F-actin deposition in the microcirculation and has various functions during host immune response. Recent reports reveal that severe COVID-19 correlates with reduced level...

Descripción completa

Detalles Bibliográficos
Autores principales: Suprewicz, Łukasz, Tran, Kiet A., Piktel, Ewelina, Fiedoruk, Krzysztof, Janmey, Paul A., Galie, Peter A., Bucki, Robert
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9694610/
https://www.ncbi.nlm.nih.gov/pubmed/36434734
http://dx.doi.org/10.1186/s12974-022-02642-4
_version_ 1784837843554664448
author Suprewicz, Łukasz
Tran, Kiet A.
Piktel, Ewelina
Fiedoruk, Krzysztof
Janmey, Paul A.
Galie, Peter A.
Bucki, Robert
author_facet Suprewicz, Łukasz
Tran, Kiet A.
Piktel, Ewelina
Fiedoruk, Krzysztof
Janmey, Paul A.
Galie, Peter A.
Bucki, Robert
author_sort Suprewicz, Łukasz
collection PubMed
description BACKGROUND: Plasma gelsolin (pGSN) is an important part of the blood actin buffer that prevents negative consequences of possible F-actin deposition in the microcirculation and has various functions during host immune response. Recent reports reveal that severe COVID-19 correlates with reduced levels of pGSN. Therefore, using an in vitro system, we investigated whether pGSN could attenuate increased permeability of the blood–brain barrier (BBB) during its exposure to the portion of the SARS-CoV-2 spike protein containing the receptor binding domain (S1 subunit). MATERIALS AND METHODS: Two- and three-dimensional models of the human BBB were constructed using the human cerebral microvascular endothelial cell line hCMEC/D3 and exposed to physiologically relevant shear stress to mimic perfusion in the central nervous system (CNS). Trans-endothelial electrical resistance (TEER) as well as immunostaining and Western blotting of tight junction (TJ) proteins assessed barrier integrity in the presence of the SARS-CoV-2 spike protein and pGSN. The IncuCyte Live Imaging system evaluated the motility of the endothelial cells. Magnetic bead-based ELISA was used to determine cytokine secretion. Additionally, quantitative real-time PCR (qRT-PCR) revealed gene expression of proteins from signaling pathways that are associated with the immune response. RESULTS: pGSN reversed S1-induced BBB permeability in both 2D and 3D BBB models in the presence of shear stress. BBB models exposed to pGSN also exhibited attenuated pro-inflammatory signaling pathways (PI3K, AKT, MAPK, NF-κB), reduced cytokine secretion (IL-6, IL-8, TNF-α), and increased expression of proteins that form intercellular TJ (ZO-1, occludin, claudin-5). CONCLUSION: Due to its anti-inflammatory and protective effects on the brain endothelium, pGSN has the potential to be an alternative therapeutic target for patients with severe SARS-CoV-2 infection, especially those suffering neurological complications of COVID-19. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02642-4.
format Online
Article
Text
id pubmed-9694610
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-96946102022-11-26 Recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the SARS-CoV-2 virus Suprewicz, Łukasz Tran, Kiet A. Piktel, Ewelina Fiedoruk, Krzysztof Janmey, Paul A. Galie, Peter A. Bucki, Robert J Neuroinflammation Research BACKGROUND: Plasma gelsolin (pGSN) is an important part of the blood actin buffer that prevents negative consequences of possible F-actin deposition in the microcirculation and has various functions during host immune response. Recent reports reveal that severe COVID-19 correlates with reduced levels of pGSN. Therefore, using an in vitro system, we investigated whether pGSN could attenuate increased permeability of the blood–brain barrier (BBB) during its exposure to the portion of the SARS-CoV-2 spike protein containing the receptor binding domain (S1 subunit). MATERIALS AND METHODS: Two- and three-dimensional models of the human BBB were constructed using the human cerebral microvascular endothelial cell line hCMEC/D3 and exposed to physiologically relevant shear stress to mimic perfusion in the central nervous system (CNS). Trans-endothelial electrical resistance (TEER) as well as immunostaining and Western blotting of tight junction (TJ) proteins assessed barrier integrity in the presence of the SARS-CoV-2 spike protein and pGSN. The IncuCyte Live Imaging system evaluated the motility of the endothelial cells. Magnetic bead-based ELISA was used to determine cytokine secretion. Additionally, quantitative real-time PCR (qRT-PCR) revealed gene expression of proteins from signaling pathways that are associated with the immune response. RESULTS: pGSN reversed S1-induced BBB permeability in both 2D and 3D BBB models in the presence of shear stress. BBB models exposed to pGSN also exhibited attenuated pro-inflammatory signaling pathways (PI3K, AKT, MAPK, NF-κB), reduced cytokine secretion (IL-6, IL-8, TNF-α), and increased expression of proteins that form intercellular TJ (ZO-1, occludin, claudin-5). CONCLUSION: Due to its anti-inflammatory and protective effects on the brain endothelium, pGSN has the potential to be an alternative therapeutic target for patients with severe SARS-CoV-2 infection, especially those suffering neurological complications of COVID-19. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02642-4. BioMed Central 2022-11-24 /pmc/articles/PMC9694610/ /pubmed/36434734 http://dx.doi.org/10.1186/s12974-022-02642-4 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Suprewicz, Łukasz
Tran, Kiet A.
Piktel, Ewelina
Fiedoruk, Krzysztof
Janmey, Paul A.
Galie, Peter A.
Bucki, Robert
Recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the SARS-CoV-2 virus
title Recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the SARS-CoV-2 virus
title_full Recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the SARS-CoV-2 virus
title_fullStr Recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the SARS-CoV-2 virus
title_full_unstemmed Recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the SARS-CoV-2 virus
title_short Recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the SARS-CoV-2 virus
title_sort recombinant human plasma gelsolin reverses increased permeability of the blood–brain barrier induced by the spike protein of the sars-cov-2 virus
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9694610/
https://www.ncbi.nlm.nih.gov/pubmed/36434734
http://dx.doi.org/10.1186/s12974-022-02642-4
work_keys_str_mv AT suprewiczłukasz recombinanthumanplasmagelsolinreversesincreasedpermeabilityofthebloodbrainbarrierinducedbythespikeproteinofthesarscov2virus
AT trankieta recombinanthumanplasmagelsolinreversesincreasedpermeabilityofthebloodbrainbarrierinducedbythespikeproteinofthesarscov2virus
AT piktelewelina recombinanthumanplasmagelsolinreversesincreasedpermeabilityofthebloodbrainbarrierinducedbythespikeproteinofthesarscov2virus
AT fiedorukkrzysztof recombinanthumanplasmagelsolinreversesincreasedpermeabilityofthebloodbrainbarrierinducedbythespikeproteinofthesarscov2virus
AT janmeypaula recombinanthumanplasmagelsolinreversesincreasedpermeabilityofthebloodbrainbarrierinducedbythespikeproteinofthesarscov2virus
AT galiepetera recombinanthumanplasmagelsolinreversesincreasedpermeabilityofthebloodbrainbarrierinducedbythespikeproteinofthesarscov2virus
AT buckirobert recombinanthumanplasmagelsolinreversesincreasedpermeabilityofthebloodbrainbarrierinducedbythespikeproteinofthesarscov2virus